Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 5 de 5
Filtrar
Mais filtros








Base de dados
Intervalo de ano de publicação
1.
Int J Mol Sci ; 14(8): 17221-37, 2013 Aug 20.
Artigo em Inglês | MEDLINE | ID: mdl-23965982

RESUMO

Periodontitis is a common inflammatory disease affecting the tooth-supporting structures. It is initiated by bacteria growing as a biofilm at the gingival margin, and communication of the biofilms differs in health and disease. The bacterial composition of periodontitis-associated biofilms has been well documented and is under continual investigation. However, the roles of several host response and inflammation driven environmental stimuli on biofilm formation is not well understood. This review article addresses the effects of environmental factors such as pH, temperature, cytokines, hormones, and oxidative stress on periodontal biofilm formation and bacterial virulence.


Assuntos
Biofilmes , Bactérias Gram-Negativas/fisiologia , Bactérias Gram-Positivas/fisiologia , Periodontite/microbiologia , Animais , Bactérias Gram-Negativas/patogenicidade , Bactérias Gram-Positivas/patogenicidade , Hemina/metabolismo , Hormônios/fisiologia , Interações Hospedeiro-Patógeno , Humanos , Concentração de Íons de Hidrogênio , Mediadores da Inflamação/metabolismo , Ferro/metabolismo , Estresse Oxidativo , Periodontite/imunologia , Periodontite/metabolismo , Virulência
2.
PLoS One ; 8(7): e70509, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23936223

RESUMO

Aggregatibacter actinomycetemcomitans is a gram-negative opportunistic oral pathogen. It is frequently associated with subgingival biofilms of both chronic and aggressive periodontitis, and the diseased sites of the periodontium exhibit increased levels of the proinflammatory mediator interleukin (IL)-1ß. Some bacterial species can alter their physiological properties as a result of sensing IL-1ß. We have recently shown that this cytokine localizes to the cytoplasm of A. actinomycetemcomitans in co-cultures with organotypic gingival mucosa. However, current knowledge about the mechanism underlying bacterial IL-1ß sensing is still limited. In this study, we characterized the interaction of A. actinomycetemcomitans total membrane protein with IL-1ß through electrophoretic mobility shift assays. The interacting protein, which we have designated bacterial interleukin receptor I (BilRI), was identified through mass spectrometry and was found to be Pasteurellaceae specific. Based on the results obtained using protein function prediction tools, this protein localizes to the outer membrane and contains a typical lipoprotein signal sequence. All six tested biofilm cultures of clinical A. actinomycetemcomitans strains expressed the protein according to phage display-derived antibody detection. Moreover, proteinase K treatment of whole A. actinomycetemcomitans cells eliminated BilRI forms that were outer membrane specific, as determined through immunoblotting. The protein was overexpressed in Escherichia coli in both the outer membrane-associated form and a soluble cytoplasmic form. When assessed using flow cytometry, the BilRI-overexpressing E. coli cells were observed to bind 2.5 times more biotinylated-IL-1ß than the control cells, as detected with avidin-FITC. Overexpression of BilRI did not cause binding of a biotinylated negative control protein. In a microplate assay, soluble BilRI bound to IL-1ß, but this binding was not specific, as a control protein for IL-1ß also interacted with BilRI. Our findings suggest that A. actinomycetemcomitans expresses an IL-1ß-binding surface-exposed lipoprotein that may be part of the bacterial IL-1ß-sensing system.


Assuntos
Aggregatibacter actinomycetemcomitans/metabolismo , Proteínas da Membrana Bacteriana Externa/metabolismo , Interleucina-1beta/metabolismo , Proteínas Recombinantes/metabolismo , Aggregatibacter actinomycetemcomitans/genética , Aggregatibacter actinomycetemcomitans/fisiologia , Sequência de Aminoácidos , Proteínas da Membrana Bacteriana Externa/genética , Proteínas da Membrana Bacteriana Externa/fisiologia , Biofilmes , Membrana Celular/metabolismo , Citosol/metabolismo , Eletroforese em Gel de Poliacrilamida , Ensaio de Desvio de Mobilidade Eletroforética , Escherichia coli/genética , Escherichia coli/metabolismo , Humanos , Immunoblotting , Dados de Sequência Molecular , Infecções por Pasteurellaceae/microbiologia , Ligação Proteica , Sinais Direcionadores de Proteínas/genética
3.
Cytokine ; 60(2): 565-74, 2012 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-22898394

RESUMO

The opportunistic pathogen Aggregatibacter actinomycetemcomitans causes periodontitis, which is a biofilm infection that destroys tooth-supportive tissues. Interleukin (IL)-1ß, a central proinflammatory cytokine of periodontitis, is an essential first line cytokine for local inflammation that modulates the cell proliferation and anti-pathogen response of human gingival keratinocytes. Previously, we demonstrated that A. actinomycetemcomitans biofilms bind IL-1ß; however, whether this binding is an active process is not known. In this study, we showed for the first time with immuno-electron microscopy that viable bacterial biofilm cells internalised IL-1ß when co-cultured with an organotypic mucosa. Decreased biofilm viability hindered the ability of biofilm to sequester IL-1ß and caused IL-1ß leakage into the culture medium. In some A. actinomycetemcomitans cells, intracellular IL-1ß localized to the outer edges of the nucleoids. We identified the DNA-binding protein HU as an IL-1ß interacting protein with mass spectroscopy and showed the interaction of recombinant HU and IL-1ßin vitro using enzyme-linked immunosorbent assay (ELISA). Close contact with a viable A. actinomycetemcomitans biofilm decreased the proliferation and apoptosis of human gingival keratinocytes as demonstrated using Ki-67 and the terminal deoxynucleotidyl transferase dUTP nick-end labelling (TUNEL) staining, respectively. Our results suggest that viable A. actinomycetemcomitans biofilms may disturb the critical first steps of local inflammation in periodontitis by binding and internalising IL-1ß. The interaction of IL-1ß with conserved HU provides a potential mechanism for shaping bacterial gene expression.


Assuntos
Aggregatibacter actinomycetemcomitans/metabolismo , Biofilmes , DNA Bacteriano/metabolismo , Endocitose , Interleucina-1beta/metabolismo , Viabilidade Microbiana , Infecções por Actinobacillus/microbiologia , Infecções por Actinobacillus/patologia , Aggregatibacter actinomycetemcomitans/efeitos dos fármacos , Aggregatibacter actinomycetemcomitans/ultraestrutura , Sequência de Aminoácidos , Apoptose/efeitos dos fármacos , Aderência Bacteriana/efeitos dos fármacos , Biofilmes/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Proteínas ELAV/química , Proteínas ELAV/metabolismo , Endocitose/efeitos dos fármacos , Células Epiteliais/efeitos dos fármacos , Células Epiteliais/microbiologia , Células Epiteliais/patologia , Gengiva/microbiologia , Gengiva/patologia , Humanos , Queratinócitos/microbiologia , Queratinócitos/patologia , Viabilidade Microbiana/efeitos dos fármacos , Dados de Sequência Molecular , Mucosa/efeitos dos fármacos , Mucosa/microbiologia , Mucosa/patologia , Mucosa/ultraestrutura , Penicilinas/farmacologia , Ligação Proteica/efeitos dos fármacos , Estreptomicina/farmacologia
4.
J Mol Biol ; 409(4): 642-53, 2011 Jun 17.
Artigo em Inglês | MEDLINE | ID: mdl-21530539

RESUMO

Secretins form large oligomeric assemblies in the membrane that control both macromolecular secretion and uptake. Several Pasteurellaceae are naturally competent for transformation, but the mechanism for DNA assimilation is largely unknown. In Haemophilus influenzae, the secretin ComE has been demonstrated to be essential for DNA uptake. In closely related Aggregatibacter actinomycetemcomitans, an opportunistic pathogen in periodontitis, the ComE homolog HofQ is believed to be the outer membrane DNA translocase. Here, we report the structure of the extra-membranous domains of HofQ at 2.3 Å resolution by X-ray crystallography. We also show that the extra-membranous domains of HofQ are capable of DNA binding. The structure reveals two secretin-like folds, the first of which is formed by means of a domain swap. The second domain displays extensive structural similarity to K homology (KH) domains, including the presence of a GxxG motif, which is essential for the nucleotide-binding function of KH domains, suggesting a possible mechanism for DNA binding by HofQ. The data indicate a direct involvement in DNA acquisition and provide insight into the molecular basis for natural competence.


Assuntos
Proteínas de Bactérias/química , DNA/metabolismo , Dobramento de Proteína , Estrutura Terciária de Proteína , Sequência de Aminoácidos , Proteínas de Bactérias/genética , Proteínas de Bactérias/metabolismo , Cristalografia por Raios X , DNA/química , Bactérias Gram-Negativas/química , Modelos Moleculares , Dados de Sequência Molecular , Multimerização Proteica , Estrutura Quaternária de Proteína , Alinhamento de Sequência
5.
PLoS One ; 6(4): e18929, 2011 Apr 18.
Artigo em Inglês | MEDLINE | ID: mdl-21533109

RESUMO

Bacterial biofilms resist host defenses and antibiotics partly because of their decreased metabolism. Some bacteria use proinflammatory cytokines, such as interleukin (IL)-1ß, as cues to promote biofilm formation and to alter virulence. Although one potential bacterial IL-1ß receptor has been identified, current knowledge of the bacterial IL-1ß sensing mechanism is limited. In chronic biofilm infection, periodontitis, Aggregatibacter actinomycetemcomitans requires tight adherence (tad)-locus to form biofilms, and tissue destroying active lesions contain more IL-1ß than inactive ones. The effect of IL-1ß on the metabolic activity of A. actinomycetemcomitans biofilm was tested using alamarBlue™. The binding of IL-1ß to A. actinomycetemcomitans cells was investigated using transmission electron microscopy and flow cytometry. To identify the proteins which interacted with IL-1ß, different protein fractions from A. actinomycetemcomitans were run in native-PAGE and blotted using biotinylated IL-1ß and avidin-HRP, and identified using mass spectroscopy. We show that although IL-1ß slightly increases the biofilm formation of A. actinomycetemcomitans, it reduces the metabolic activity of the biofilm. A similar reduction was observed with all tad-locus mutants except the secretin mutant, although all tested mutant strains as well as wild type strains bound IL-1ß. Our results suggest that IL-1ß might be transported into the A. actinomycetemcomitans cells, and the trimeric form of intracellular ATP synthase subunit ß interacted with IL-1ß, possibly explaining the decreased metabolic activity. Because ATP synthase is highly conserved, it might universally enhance biofilm resistance to host defense by binding IL-1ß during inflammation.


Assuntos
Proteínas de Bactérias/metabolismo , Biopolímeros/metabolismo , Interleucina-1beta/metabolismo , Pasteurellaceae/enzimologia , Biofilmes , Citometria de Fluxo , Humanos , Microscopia Eletrônica de Transmissão , Ligação Proteica
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA